You are on page 1of 9

Biomedicine & Pharmacotherapy 142 (2021) 112021

Contents lists available at ScienceDirect

Biomedicine & Pharmacotherapy


journal homepage: www.elsevier.com/locate/biopha

Hidden pharmacological activities of valproic acid: A new insight


Dhirendra Singh a, Sumeet Gupta a, *, Inderjeet Verma a, Mohamed A. Morsy b, c, Anroop B. Nair b,
Al-Shaimaa F. Ahmed d
a
Department of Pharmacology, M.M. College of Pharmacy, M.M. (Deemed to be University), Mullana, Ambala, Haryana, India
b
Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa, Saudi Arabia
c
Department of Pharmacology, Faculty of Medicine, Minia University, El-Minia, Egypt
d
Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, El-Minia, Egypt

A R T I C L E I N F O A B S T R A C T

Keywords: Valproic acid (VPA) is an approved drug for managing epileptic seizures, bipolar disorders, and migraine. VPA
Valproic acid has been shown to elevate the level of gamma-aminobutyric acid (GABA) in the brain through competitive in­
Histone deacetylases hibition of GABA transaminase, thus promoting the availability of synaptic GABA and facilitating GABA-
Diabetes mellitus
mediated responses. VPA, which is a small chain of fatty acids, prevents histone deacetylases (HDACs).
Renal protection
Cardiac remodeling
HDACs play a crucial role in chromatin remodeling and gene expression through posttranslational changes of
Muscular dystrophy chromatin-associated histones. Recent studies reported a possible effect of VPA against particular types of
cancers. This effect was partially attributed to its role in regulating epigenetic modifications through the inhi­
bition of HDACs, which affect the expression of genes associated with cell cycle control, cellular differentiation,
and apoptosis. In this review, we summarize the current information on the actions of VPA in diseases such as
diabetes mellitus, kidney disorders, neurodegenerative diseases, muscular dystrophy, and cardiovascular
disorders.

1. Introduction channels [8]. Moreover, an effect of VPA, which was attributed to its
action on histone deacetylase (HDAC), was reported against leukemia
Valproic acid (VPA), commonly known as valproate, is among the and a few solid tumors. VPA was also shown to exhibit cell-specific
most commonly prescribed antiepileptic drugs [1]. Globally, approxi­ selectivity by attenuating HDAC6 and HDAC8 activity in a model of
mately one million people consume VPA daily [2]. VPA is an analog of cardiac hypertrophy [9]. Collectively, it is clear that VPA exerts multiple
valeric acid (a natural product obtained from Valeriana officinalis, syn­ actions through different mechanisms; thus, the review aims to highlight
thesized by Beverly S. Burton in 1882 to be used as a solvent [3]). The and discuss its new pharmacological actions.
antiepileptic action of VPA was discovered in 1962 by Pierre Eymard
[4]. In 1967, VPA was officially approved in France as an antiepileptic 2. Actions on the CNS
agent. It is currently approved in more than one hundred countries [5].
VPA is also used for other indications including, migraine headache, VPA, an FDA-approved antiepileptic drug, is a propyl substituent on
bipolar disorder, and as a protective agent against diabetic neuropathy a pentanoic acid stem [10]. It is the drug of choice in treating general­
[6]. ized or partial epilepsies, primarily for the management of absence
VPA acts by potentiating the effect of the inhibitory transmitter seizures [11]. It modulates neuronal discharge by enhancing GABA
gamma-aminobutyric acid (GABA) via prevention of GABA degradation, levels in pre and postsynaptic neurons. The binding of GABA to its own
suppression of GABA transamination action, and promotion of GABA receptor leads to chloride ion influx into the postsynaptic neuron,
synthesis [7]. Additionally, VPA prolongs the N-methyl-­ leading to hyperpolarization of the neurons [12]. VPA enhances GABA
D-aspartate-mediated stimulation with inhibition of calcium and sodium action by enhancing its synthesis by increasing glutamic acid

Abbreviations: CHOP, lowered C/EBP-homologous protein; GABA, gamma-aminobutyric acid; GRP78, glucose-regulated protein 78; HDACs, histone deacetylases;
HSP, heat shock protein; ROS, reactive oxygen species; T1DM, type 1 diabetes mellitus; VPA, valproic acid.
* Corresponding author.
E-mail address: sumeetgupta25@gmail.com (S. Gupta).

https://doi.org/10.1016/j.biopha.2021.112021
Received 4 April 2021; Received in revised form 28 July 2021; Accepted 7 August 2021
Available online 25 August 2021
0753-3322/© 2021 The Author. Published by Elsevier Masson SAS. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/4.0/).
D. Singh et al. Biomedicine & Pharmacotherapy 142 (2021) 112021

decarboxylase, an enzyme involved in the biosynthesis of GABA, and by connective tissue growth factor, and transforming growth factor-β1
delaying its metabolism through the suppression of GABA transaminase [22]. VPA has been shown to attenuate cardiac hypertrophy and fibrosis
and succinic semialdehyde dehydrogenase, which metabolize GABA. by inhibiting HDACs to acetylate the mineralocorticoid receptor in
Moreover, VPA enhances the expression of GABA receptors, therefore spontaneously hypertensive rats [23].
amplifying the neuronal response to GABA [13]. At a dose of 300 mg/kg once daily for 4 weeks, VPA suppressed the
sympathetic excitability, reduced cardiac remodeling, and modified left
3. VPA as an HDAC inhibitor ventricular work during a pressure overload experimental study
demonstrated by Yang et al. [24]. In transgenic mice, 8 weeks of VPA
VPA, as well as other HDAC inhibitors, can alter the expression of administration attenuated atrial remodeling and slowed down atrial
many genes [14]. Corresponding proteins were described to play fibrillation. This was attributed to a marked reduction in (a) atrial
important roles in cellular activity and could influence several important dilation and fibrosis, (b) enlargement of cardiomyocytes, and (c)
pathways such as cell cycle regulation, differentiation, apoptosis, as well disarrangement in myocytes’ structure via regulating oxidative phos­
as DNA repair [15]. Thus, multiple actions were reported in different phorylation or RhoA signaling [25].
research studies. For example, it reduced the action of HDAC6 and Tian and his colleagues reported that VPA in a dose of 250 mg/kg in
HDAC8 in a cardiac hypertrophy model [9]. In renal fibrosis, VPA rats showed a 50% reduction in myocardial infarct volume and
inhibited the effect of HDAC4/5 [16]. It also suppressed the activity of improved systolic blood pressure via HDAC inhibition and Foxm1 gene
HDAC1/2 in hepatic fibrosis [17] and HDAC3/4 activity in a penile expression with improved survival of cardiomyocytes cells [26].
fibrosis model [18]. HDAC suppression correlates with beneficial effects Another study showed a hypotension effect via minimizing the HDAC1
for several neuronal diseases because class I and II HDACs remarkably expression and reducing angiotensin II in a high-fat-diet rat model after
affect neuronal activity [4]. VPA can alter DNA methylation by the ac­ 6 days of VPA treatment at a dose of 300 mg/kg [27]. Moreover, VPA
tion of DNA methyltransferases, which add a methyl moiety through blocked sepsis-mediated myocardial dysfunction by accelerating
S-adenosyl-L-methionine to the 5th carbon of CpG dinucleotide cyto­ myocardial autophagy through enhancing the expression of PTEN and
sine, which leads to silencing of the gene [19]. VPA was shown to lower suppressing the AKT/mTOR pathway [28].
methylated DNA in many cell lines such as neuroblastoma, HeLa, and
HEK 293. Moreover, mice treated with VPA displayed elevated deme­ 5. Action on diabetes mellitus
thylation of DNA in their brains [20].
Diabetes mellitus (DM) is a metabolic disorder characterized by
4. Actions on the cardiovascular system chronic hyperglycemia with abnormal carbohydrate, fat, and protein
metabolism due to defects in insulin secretion, insulin action, or both.
VPA showed a cardioprotective action (Fig. 1) through its action on It is classified into type 1 diabetes mellitus (T1DM) as well as T2DM
HDACs and its ability to affect genetic regulation [21]. Li et al. studied [29]. T1DM is mainly caused by autoimmune β-cell damage causing
the cardioprotective action on a rat renovascular hypertensive model. endogenous insulin deficiency [30]. Autoimmune antibodies that work
VPA (400 mg/kg) ameliorated cardiac dysfunction, cardiac hypertro­ against insulinoma antigen II, glutamic acid decarboxylase, insulin, and
phy, and fibrosis by suppressing the expression of HDAC2, HDAC8, the recently characterized zinc transporter VIII, are among the most

Fig. 1. The therapeutic potential of valproic acid (VPA) in cardiac dysfunction. VPA inhibits the activity of histone deacetylases (HDACs), which leads to histone
acetylation and relaxation of chromatin structure, resulting in gene expression and shows beneficial outcomes for cardiac arrhythmia, cardiac fibrosis, cardiac
hypertrophy, hypertension and myocardial infarction.

2
D. Singh et al. Biomedicine & Pharmacotherapy 142 (2021) 112021

efficient biological markers for autoimmune T1DM in both adults and 7. Anti-inflammatory effect of VPA
children [31].
In a daily oral dose of 150 and 300 mg/kg for 3 weeks, VPA improved Recent research demonstrated the anti-inflammatory action of VPA
glycemic control by suppressing HDAC activity and enhancing the based on a specific molecular and cellular level in post-surgical
number of β-cells and their function in the streptozotocin-induced rat inflammation of the conjunctiva, where a mouse conjunctival scarring
diabetic model [32]. Additionally, VPA also reduced insulin resistance, model was used to evaluate the effect of VPA. After surgery, VPA was
fat accumulation, dyslipidemia, and hepatic glucose production in injected immediately into the sub conjunctiva. It suppressed the gen­
T2DM, suggesting its effects on peripheral tissues like liver and adipose eration of pro-inflammatory cytokines such as CXCL1, IL5, and IL6, the
tissue [33]. Moreover, studies conducted on Wfs1-knockout mice and reduction of IL10, and the TNFα-induced NF-κB [45]. Liu and his col­
streptozotocin-injected mice showed a reduction in blood glucose level leagues explored the protective effect of VPA in a rat model of autoim­
after administering 300 mg/kg of VPA via insulin enhancing action mune encephalomyelitis. VPA reduced inflammatory responses,
[34]. Another study conducted on alloxan diabetic rats treated with VPA minimized microgliosis, repressed activation of NF-κB signal pathway,
reported glucose-lowering, antihyperlipidemic, and antioxidant effects and attenuated retinal ganglion cell apoptosis in rats’ optic nerve [46].
[35]. A similar effect was noted in a different study conducted on a VPA minimized the nociception and inflammation in a rodent model of
high-fat diet/streptozotocin-treated rats [36]. acute pain and inflammation. The effect of VPA was examined on
carrageenan-induced paw edema and peritonitis. VPA treatment
6. Renal protective effect of VPA reduced paw edema and protected tissue structure. Reduced levels of
TNFα and leukocytes and releases of myeloperoxidase in peritonitis
When there is an imbalance between the production of free radicals were also observed [47].
and antioxidants, oxidative stress occurs. Consequently, individual
biomolecules are oxidized, and these molecules are structurally and 8. Neuroprotective effect of VPA
functionally modified [37]. This oxidizing process is mainly conducted
in the mitochondria, using mitochondrial cytochrome oxidase enzymes Neural network rhythms are engaged in information processing,
like cytochrome P450. Reactive oxygen species (ROS) products from storage, and retrieval, which can be vital for memory consolidation,
such a mechanism probably lead to progressive kidney damage and executive functioning, and sensory processing. Thus, brain arrhythmias
atherosclerotic disease resulting in chronic renal disease [38]. might have a catastrophic effect on circuit operation, which is consid­
In a model of diabetic nephropathy, Sun et al. found that VPA ered the initial symptom of numerous neurological diseases. Further­
attenuated diabetes-mediated renal injury through suppressing endo­ more, brain arrhythmia can function as biomarkers for a broad area of
plasmic reticulum-induced stress and apoptosis. VPA enhanced the level brain disease [48]. Imbalance in acetylation of histone and, as a result,
of glucose-regulated protein 78 (GRP78) and lowered C/EBP- the malfunction in transcription is related to a large variety of neuro­
homologous protein (CHOP), gene 153, and caspase‑12 levels. This degenerative diseases (Fig. 2) [49].
result might be attributed to histone H4 acetylation regulation in the VPA has been shown to enhance white matter renovation and the
promoter site of the GRP78 and CHOP in the endoplasmic reticulum formation of neurons after stroke. Administration of VPA 28 days
[39]. Additionally, treatment with VPA had a beneficial effect on following middle cerebral artery occlusion (an experimental stroke
podocyte damage caused by adriamycin in mice by blocking flagellation model) dramatically improved survival of oligodendrocyte and facili­
and detachment of the podocyte foot process, as well as the glomer­ tated oligodendrogenesis, resulting in an increased myelinated axon
ulosclerosis process [40]. Furthermore, Khan et al. confirmed the density in the ischemic penumbra [50]. VPA was reported to attenuate
renoprotective actions of VPA through its ability to reduce podocyte and brain injury in a rat transient focal cerebral ischemia model by inhibiting
renal injuries, specifically by promoting the autophagy process and HDAC and inducing the level of heat shock protein (HSP). VPA treat­
deactivation of NF-κB/inducible nitric oxide synthase signal pathway in ment (300 mg/kg) right after ischemia followed by repetitive injections
a rat diabetic nephropathy model [16]. every 12 h was noticed to diminish infarct size and lower
Speir et al. demonstrated the beneficial effect of VPA against acute ischemia-induced neurological insufficiency scores. It also inhibited the
renal ischemia-reperfusion damage in the rat. VPA at a dose of 150 mg/ stimulation of caspase-3 [51].
kg minimized ischemic injury by downregulation of inflammatory cy­ Wang et al. showed that chronic VPA therapy stimulated angiogen­
tokines IL1β and IL6 and upregulation of anti-apoptotic molecule Bcl2 esis and facilitated functional regeneration after rat ischemic stroke in
and improved renal tissue regeneration [41]. In acute kidney injury the brain. They explained these observations by the ability of VPA to
induced by sepsis in mice, VPA (300 mg/kg) effectively ameliorated suppress HDAC and activate hypoxia-inducible factor-1α and its asso­
kidney damage by minimizing oxidative and inflammatory responses. ciated proangiogenic vascular endothelial growth factor and matrix
VPA treatment reduced renal malondialdehyde and myeloperoxidase metalloproteinase2/9 [52]. A recent study also proved that
activity while enhancing the level of superoxide dismutase and the VPA-induced suppression of glycogen synthase kinase-3 and HDAC is
reduced form of glutathione along with minimization in circulating responsible for its neuroprotective effect in a rat brain ischemia model
concentrations of TNFα, IL1β, and IL6 [41]. [53]. Moreover, VPA showed a neuroprotective effect against rat global
Epigenetic gene modifications are strongly regulated by histone transient ischemia model by attenuating hippocampal cell loss and
deacetylation [42]. The intricate connection between inhibitors of cognitive deficits via reducing ROS production and cerebral inflamma­
HDAC action in the epigenetic maintenance and modification of immune tion and repression of HDAC and activation of HSP70 [54].
cell gene expression, particularly concerning myeloid cell and T Ximenes et al. have demonstrated the neuroprotective action of VPA
lymphocyte differentiation and activation, has been well recognized in the 6-OHDA rat model of Parkinson’s disease. In their study, they
[43]. Amirzargar et al. have noticed that VPA changed the cytokine showed that intranigral administration of 6-OHDA to rats enhanced the
profile and displayed anti-inflammatory action in a rat renal release of IL1β, IL6, and TNFα in the striatum, while oral administration
ischemia-reperfusion damage model. Treatment with VPA minimized of VPA at 50 mg/kg reduced the level of inflammatory mediators and
histopathological alteration and serum creatinine level, attenuated the decreased neuron degeneration in the striatum owing to the anti-
response of inflammatory mediators, and enhanced the inflammatory and HDAC inhibitory actions of VPA [55]. Moreover,
anti-inflammatory state to support quicker renal recovery due to VPA displayed a neuroprotective action against rotenone-induced Par­
ischemic alteration and better improvement of kidney function [44]. kinson’s disease. Administration of the mitochondrial toxin, rotenone,
induced injury in dopaminergic neurons while chronic treatment of VPA
reduced the nuclear translocation of synuclein monoubiquitinated

3
D. Singh et al. Biomedicine & Pharmacotherapy 142 (2021) 112021

Fig. 2. The beneficial prospects of valproic acid (VPA)


in neurodegenerative disease. VPA blocks the action
of histone deacetylases (HDACs) and leads to tran­
scriptional activation of anti-apoptotic and neuronal
surviving pathways like HSP70, AKT, CDK5, ERK, NF-
κB, and suppression of caspase-3, TNF-α, IL-1β, IL-6,
ROS, and many other factors which cause neuronal
death. HSP70: Heat Shock Protein 70; AKT: Enzyme
serine/threonine protein kinase; CDK5: Cyclin
Dependent Kinase 5; ERK: Extracellular Signal-
regulated Kinases; TNF-α: Tumor Necrosis Factor-α;
IL-1β: Interleukin-1β; ROS: Reactive Oxygen Species;
NF-κB: Nuclear Factor-κB; MMP: Matrix metal­
loproteinases; GSK: Glycogen Synthase Kinase.

protein in dopaminergic neurons and protected substantia nigra neu­ decreased Aβ production, reduced neuritic plaque formation, and alle­
rons, and increased the level of tyrosine hydroxylase and striatal dopa­ viated the memory deficits in AD transgenic mice [60]. VPA improved
mine level. By enhancing histone H3 acetylation and inhibiting the the survival rate and abnormal motor activity in a model of Huntington’s
HDAC enzyme, VPA was able to increase the expression of neuro­ disease. Daily administration of VPA increased the survival of transgenic
protective genes [56]. mice and restored their reduced involuntary locomotor performance
VPA attenuated neuron injury and Parkinson’s disease-like charac­ without showing any prominent adverse effect on their behavior [61].
ters in LRRK2 R1441G transgenic mice. VPA treatment improved his­ VPA administration alleviated traumatic brain injury (TBI)-induced
tone acetylation levels and the number of tyrosine hydroxylase- blockade of autophagic flux and increased the autophagy stimulation,
containing neurons in substantia nigra of mice. It also decreased iba-1- thus inhibiting apoptosis of brain cells and affecting microglial activa­
stimulated microglia and the level of pro-inflammatory mediators, tion and phenotype polarization to promote post-traumatic functional
thus improving parkinsonism-associated motor and non-motor deficits recovery and serve as a possible post-traumatic brain injury treatment
[57]. In vitro, VPA protected dopaminergic SH-SY5Y cells from [62].
MPP+-produced neurotoxicity. Pretreatment with VPA attenuated Similarly, another study reported that VPA exerted anti-apoptotic
MPP+-induced dopaminergic cell death by inhibiting ROS production and neuroprotective effects against TBI by up-regulating ERK and AKT
via activating CDK5/p35 cascade and ERK signaling pathway [58]. Leng expression, and these signaling pathways mediate VPA neuroprotection
et al. suggested that VPA could induce α-synuclein in rat cerebral [63]. VPA showed anti-apoptotic and neuroprotection qualities in
cortical neurons both in vitro and in vivo via inhibition of HDAC, which SH-SY5Y cells and primary cortical neurons by inhibiting endoplasmic
contributed to its neuroprotection against glutamate excitotoxicity [59]. reticulum stress and promoting the AKT activity.
VPA inhibited GSK-3β-mediated γ-secretase cleavage of APP, Hernandez and his team have revealed the neuroprotective action of

4
D. Singh et al. Biomedicine & Pharmacotherapy 142 (2021) 112021

VPA against ischemia-reperfusion injury in the brain of rats. VPA important role in cancer research [74]. In MCF-7 breast carcinoma cells,
treatment alters Bcl2 expression and inhibits caspase-3 activation, VPA enhanced the antiproliferative effect of melatonin by inducing the
which may be a protective mechanism of VPA against apoptosis in expression of melatonin receptor MT1 [75]. Terranova et al. found that
injured neurons [64]. Moreover, VPA was also reported to reduce VPA at low concentration produced both time and dose-dependent
apoptosis through up regulation of Bcl2 and down regulation of Bax upregulation of thymidine phosphorylase transcript and protein
expression in developing neurons in a study in which VPA was given to expression in breast carcinoma cells. However, this effect was not
pregnant rats [65]. This effect is indeed explaining the toxic effects of recorded in the non-cancer breast MCF-10 cell line. The authors also
VPA (teratogenicity) after fetal exposure. found that VPA potentiated capecitabine action in breast carcinoma cells
by inhibiting HDAC3; however, this effect was lost in thymidine phos­
9. Effect of VPA on drug addiction phorylase knockout cells [76].
VPA also showed a synergistic anticancer action with arsenic trioxide
Regulation of chromatin arrangement during post-translation alter­ against NCI-H1299 and NCI-H460 lung cancer cells both in vitro and in
ations of histones acetylation shows up as an essential mechanism to vivo. This combination efficiently suppressed the proliferation of lung
transform a diversity of environmental stimuli involving drugs of abuse cancer cells by arresting the G2/M-phase of the cell cycle and inducing
into specific modification in genetic expression. Changes in the expres­ caspase-dependent apoptosis [77]. Additionally, the combined syner­
sion of genes support the occurrence and continuation of the addicted gistic action of VPA and gemcitabine was investigated on pancreatic
condition [66]. A study carried out on mice demonstrated that repeated tumor cells. It was observed that VPA dose-dependently improved the
injections of VPA after methamphetamine might minimize the addictive sensitivity of gemcitabine towards pancreatic tumor cells. Surprisingly,
behavior. On the other hand, this result was not observed on a low VPA concentration augmented the invasion of pancreatic carci­
cocaine-caused behavior. Such primary outcome displayed that different noma cells that previously displayed gemcitabine-stimulated motility.
abuse drugs have different neural mechanisms, which indicate that VPA Furthermore, a low VPA dose enhanced the ROS/AKT/STAT3/Bmi1
may possess distinct modulating action on methamphetamine and pathway activation and eventually supported the migration and tissue
cocaine addiction in humans [67]. invasion of pancreatic tumors caused by gemcitabine [78]. Similarly, a
A non-randomized and open-label clinical study has been conducted study evaluated the effect of combining VPA with P276-00, a
to compare the short-term effect of buprenorphine and VPA to a control cyclin-dependent kinase blocker, on lung cell carcinoma. The results of
group that got a 10-day conventional treatment of clonidine/carba­ this study revealed that this combination is synergistic whether tested on
mazepine. Sixty-two-drug dependent subjects were included to evaluate p53+ or p53- lung cell carcinoma with a significant decrease in colony
the efficacy of VPA and buprenorphine against drug dependence. The formation, up-regulation of tumor inhibitor proteins such as p53, p27,
combined treatment of buprenorphine and VPA appeared to be a and p21, and a reduction of the expression of survival proteins like Bcl2
harmless and hopeful method for therapy of several drug withdrawal and cyclin D1 [79].
symptoms because the combination of both drugs did not affect blood VPA was investigated for anticancer activity (Fig. 3) on the FaO cell
pressure, pulse, or liver function and displayed no drug interaction and line of rat hepatoma. It induced the death of rat hepatoma cells through
having few side effects. These outcomes indicate that a combination of apoptosis in a dose-dependent way. Upregulation of caspase-11 was
buprenorphine/VPA may be a better de-addiction therapy for with­ enhanced at the transcription level and caspase-3 at the enzyme activity
drawal symptoms of many drugs than the conventional management level, proposing a caspase role in VPA-mediated apoptosis in FaO cells
with clonidine and carbamazepine [68]. [80]. In prostate cancer, the combined effect of metformin and VPA
produced more apoptosis and inhibition of prostate PC-3 and LNCaP
10. Antimicrobial activity of VPA cancer cells due to activation of the p53 signaling pathway [81]. VPA
was also found to exert a synergistic effect with metformin in
Microbial resistance to antibiotics represents a major public health metformin-resistant renal cancer cells. This effect was attributed to the
problem, particularly in developing countries where infectious diseases ability of VPA to enhance H3 acetylation [82]. In the thyroid cancer cell
still represent a serious cause of human mortality [69]. Antibiotics are line, the anticancer action of VPA was examined on cancer growth and
either cytotoxic or cytostatic to pathogenic microorganisms, helping the Notch1 upregulation. VPA showed an ability to activate Notch1
body’s normal defenses to eradicate them [70]. VPA displayed a expression, stimulate apoptosis, and suppress cancer cell growth [83].
powerful antifungal effect in a pH-dependent manner towards sensitive Michaelis et al. [84] studied the effects of VPA combined with IFN-α
and resistant Candida albicans. VPA exerted a potent anti-biofilm effect against two neuroblastoma cell lines (NB-2 and UKF-NB-3). They found
and decreased vaginal epithelium cell damage induced by Candida that VPA and IFN-α inhibited the growth of UKF-NB-3 xenograft tumors
albicans and potentiated the effect of antifungals such as terbinafine. The of nude mice synergistically and promoted complete healing in two out
antifungal activity of VPA was attributed to its ability to interfere with of six animals, whereas single therapy only repressed tumor growth. In
the fungal vacuoles’ integrity [71]. In a study by Nathiya et al., the gastric tumor cells, VPA administration suppressed HDAC1/2 action and
antibacterial action of several anticonvulsant drugs, including VPA, was produced enhanced autophagy, which leads to stimulation of apoptosis
tested. VPA displayed high effectiveness against Staphylococcus aureus through repression of the inhibition of HDAC1/PTEN/AKT signal
and Proteus vulgaris at 100 µg/ml, while other tested organisms required transduction pathway [85]. Li and Ma studied the adjuvant effect of VPA
more than 200 µg/ml. Carbamazepine and gabapentin were not effec­ and a diterpenoid oridonin to suppress the proliferation and metastasis
tive against these isolated bacteria. Therefore, it was concluded that formation of HL-60 leukemia cells. Oridonin showed apoptotic action on
VPA, among other anticonvulsants, has a unique antibacterial action and HL-60 leukemia cells by stimulating the intrinsic apoptotic pathways,
might be useful against certain bacterial infections [72]. while the drug combination also downregulated Bcl2/Bax ratio [86].
VPA also time and dose-dependently enhanced the effect of GANT61
11. Anticancer activity in suppressing the growth of multiple myeloma cells by blocking the
upregulated PTCH1, GLI1, and HES-1 signaling pathway [87]. Inter­
Histone deacetylation and DNA methylation are mechanisms estingly, the anticancer effect of VPA was not only shown in experi­
involved in resistance to chemotherapy through the protection of the mental studies but was also included in clinical trials. For example, VPA
cancerous cells via silencing of crucial anti-tumor genes. It is supposed in combination with karenitecin, a topoisomerase I inhibitor, underwent
that HDAC inhibitors function via stimulation or de-repression of phase I/II clinical study where 47% of melanoma patients on combi­
silenced tumor inhibitor genes [73,74]. HDACs possess a vital impact on nation therapy possessed a stable disease with an average
chromatin remodeling and epigenetics, giving their inhibitors an progression-free longevity of 10.3 weeks, while 34% of patients on

5
D. Singh et al. Biomedicine & Pharmacotherapy 142 (2021) 112021

Fig. 3. The favorable role of valproic acid


(VPA) in cancer. VPA has an important impact
on cancer prevalence by activating p53, PTEN,
Bax, caspase-3, ROS, p21, and p27, which leads
to cancer cell cycle arrest, growth, and
apoptosis. VPA also inhibits cyclin D1, AKT,
STAT, TNF-α, NF-κB, and Bcl2, which cause
cancer survival by blocking the apoptosis
pathway. PTEN: Phosphatase and Tensin ho­
molog deleted on chromosome 10; Bax: Bcl2
associated X; caspase-3: cysteine aspartic
protease-3; ROS: Reactive Oxygen Species;
AKT: Enzyme serine/threonine protein kinase;
TNF-α: Tumor Necrosis Factor-α; STAT: Signal
Transducers and Activators Transcription; NF-
κB: Nuclear Factor-κB; Bcl2: B cell lymphoma 2.

single therapy had stable disease with an average progression-free fibrosis, hind limb spasms, improved sarcolemmal integrity, and
longevity of 7.9 weeks [88]. VPA also increased the sensitivity of can­ reduced CD8+ inflammatory cells in Duchenne muscular atrophy mice.
cer cells to chemotherapy, produced growth arrest, induced the Moreover, VPA displayed protective effects on the myotubes of skeleton
apoptosis of glioma cells, and improved the survival rate of patients in a muscle, activated AKT signaling, enhanced gene transcription and pro­
clinical trial when given in combination with one or more chemother­ tein synthesis, and promoted cell longevity by inhibiting apoptosis [97,
apeutic agents with advanced melanoma [89]. 98]. Additionally, VPA was found to improve muscle wasting produced
In one clinical trial on patients with acute myeloid leukemia, 11 out during cancer cachexia in mice by inhibiting C/EBPβ binding to
of 36 patients (average age 77) unsuitable for normal intensive ubiquitin-ligase MAFbx/atrogin promoter and enhancing protein anab­
chemotherapy showed response to a combination of VPA, all-trans ret­ olism by inducing AKT phosphorylation [99]. A phase II open-label trial
inoic acid, and a lower dose of cytarabine. It was noted that the drug of VPA concluded that the drug might be used safely in patients with
combination was associated with a low frequency of adverse effects and spinal muscular atrophy older than 2 years. There was some evidence in
complete remission in two patients [90]. A study conducted in 2013 support of improvement in motor function in younger non-ambulatory
indicated that VPA could reduce the maximum tolerated dose of tem­ children [100].
sirolimus in pediatric patients with solid tumors. The combined treat­
ment was well tolerated and maintained the blocking effect of 13. Other activities
temsirolimus with a minimal degree of toxicity on normal CD34+ he­
matopoietic precursors, confirming their valuable and safe role in 13.1. Effect on eyes
treating B lymphoma [91].
Maintained VPA trough concentrations of 75–100 μg/ml in children In rat models of retinal ischemia/reperfusion injury induced by
with refractory solid tumors in phase I study was well-tolerated. increasing intraocular pressure, pretreatment with 300 mg/kg subcu­
Hyperacetylation of histone in peripheral blood mononuclear cells was taneous VPA reduced retinal damage [101]. Subsequent studies
recorded in half of the studied patients [92]. A phase II clinical study was demonstrated that VPA attenuates retinal injury by promoting antioxi­
conducted to assess the advantages of adding VPA to weekly paclitaxel dant effects, anti-apoptotic pathways via histone H3 acetylation, and the
in patients with gastric cancer. In this study, no significant advantage up-regulation of HSP70 [102,103].
was observed between the weekly paclitaxel (median overall survival
9.8 months) and the weekly paclitaxel plus VPA (median overall survival
13.2. Effect on lungs
8.7 months) [93]. VPA reduced the frequency of myeloid-derived sup­
pressor cells, accompanied by the down-regulation of TLR4 mRNA
In a rat model of acute lung injury induced by superior mesenteric
expression [94].
artery occlusion, VPA has been shown to improve survival and mitigate
lung injury through ROS reduction and anti-inflammatory effects [104].
12. Protective role of VPA in muscular dystrophy
VPA treatment also provided pulmonary protection by expressing the
antioxidant heme oxygenase 1 after ischemia/reperfusion injury [105].
Muscular dystrophies consist of a distinct class of genetic disorders,
which results in muscle degeneration with impaired function [95].
HDAC can control gene transcription in muscular progenitor cells via 13.3. Effect on muscles
managing the action of myogenic regulatory factors and myocyte
enhancer factor-2 family proteins [96]. VPA administration reduced VPA has been found to alleviate muscle wasting in rodents’ models of
cancer cachexia and myotubes exposed to conditioned medium from

6
D. Singh et al. Biomedicine & Pharmacotherapy 142 (2021) 112021

cachexia-inducing cell lines. Notably, VPA prevented an up-regulation ICAM-1, suggesting its potential as a therapeutic agent during Covid-19
of muscle C/EPBβ expression in these models [99, 106–108]. infection [115]. A short open-label clinical trial was conducted on
Covid-19 patients who showed promising effects on fatality and hospital
13.4. Effect on hormones stay length [116].

Lagace et al. demonstrated that VPA decreases in a dose-dependent Funding


manner leptin secretion and leptin mRNA levels in adipocytes in vitro,
suggesting that VPA therapy may be associated with altered leptin ho­ No funding was received from any source.
meostasis contributing to weight gain in vivo [109].
Author Contributions
13.5. In silico studies
Conceptualization, Dhirendra Singh and Sumeet Gupta; literature
Elbadawi and associates reported a possible activity of VPA against review, Dhirendra Singh, Sumeet Gupta, Mohamed A. Morsy, Anroop B.
Plasmodium falciparum HDAC1. They proposed that VPA would also Nair and Al-Shaimaa F. Ahmed; writing–original draft preparation,
have anti-leishmanial activity mediated by inhibiting Leishmania dono­ Dhirendra Singh and Sumeet Gupta; Writing–review & editing, Sumeet
vani class I HDAC [110]. In silico docking study reported that Gupta, Mohamed A. Morsy, Anroop B. Nair and Al-Shaimaa F. Ahmed.
VPA-induced inhibition to GABA transaminase and succinate semi­ All authors read and approved the final manuscript.
aldehyde dehydrogenase are mainly responsible for up-regulating GABA
expression to play an anticonvulsant effect [111].
Conflict of interest statement
14. Conclusion
The authors report no declarations of interest.
VPA was brought up to clinical practice 5 decades ago by chance.
Today, VPA is utilized to treat many diseases, and its tolerability and References
utility across ailments are well established. Information from many
[1] E. Perucca, Pharmacological and therapeutic properties of valproate, CNS Drugs
preclinical and clinical trials indicates that VPA might have actions
16 (10) (2002) 695–714.
mediated through molecular mechanisms that are yet unknown. Besides [2] E. Farinelli, D. Giampaoli, A. Cenciarini, E. Cercado, A. Verrotti, Valproic acid
the advancement of innovative antidiabetic, neuroprotective, car­ and nonalcoholic fatty liver disease: a possible association? World J. Hepatol. 7
(9) (2015) 1251–1257.
dioprotective along with other healing substances with HDAC inhibiting
[3] B.S. Burton, On the propyl derivatives and decomposition products of
properties, well-designed randomized control trials are required to ethylacetoacetate, Am. Chem. J. (1882).
verify the effectiveness of VPA as adjuvant therapy for various disorders. [4] S. Chateauvieux, F. Morceau, M. Dicato, M. Diederich, Molecular and therapeutic
Data on pharmacokinetics, half-life, oral bioavailability, and adverse potential and toxicity of valproic acid, J. Biomed. Biotechnol. 2010 (2010) 2010.
[5] R.H.M.R. Levy, B.S. Meldrum, E. Perrucca (Eds.), Antiepileptic Drugs, fifth ed,
reactions of VPA during chronic therapy are available. Lippincott Williams Wilkins, Philadelphia, 2002.
Interestingly, the VPA has both pro-apoptotic and anti-apoptotic [6] V. Bril, J. England, G.M. Franklin, M. Backonja, J. Cohen, D. Del Toro,
properties. In several neuroprotective studies, VPA showed anti- E. Feldman, D.J. Iverson, B. Perkins, J.W. Russell, D. Zochodne, N. American
Academy of, M. American Association of Neuromuscular and Electrodiagnostic,
apoptotic effects by activation of Bcl2, HSP70, CDK5, ERK, AKT R. American Academy of Physical Medicine and, Evidence-based guideline:
pathway and inhibition of caspase-3, Bax, ROS, and endoplasmic retic­ treatment of painful diabetic neuropathy: report of the American Academy of
ulum stress, whereas in anticancer studies, VPA exerted its action Neurology, the American Association of Neuromuscular and Electrodiagnostic
Medicine, and the American Academy of Physical Medicine and Rehabilitation,
mechanism through activation of caspase-3, Bax, p21, endoplasmic re­ Pm&r 3 (4) (2011) 345–352, e21.
ticulum stress and ROS formation and inhibition of Bcl2, cyclin D1, and [7] M.F. Silva, C.C. Aires, P.B. Luis, J.P. Ruiter, L. IJlst, M. Duran, R.J. Wanders,
AKT. This may be due to different signaling mechanisms between I. Tavares de Almeida, Valproic acid metabolism and its effects on mitochondrial
fatty acid oxidation: a review, J. Inherit. Metab. Dis. 31 (2) (2008) 205–216.
normal and cancer cells.
[8] A. VanDongen, M. VanErp, R. Voskuyl, Valproate r0educes excitability by
Several publications introduced the beneficial actions of VPA and its blockage of sodium and potassium conductance, Epilepsia 27 (3) (1986)
potential side effects such as hepatotoxicity, aplastic anemia, coagulo­ 177–182.
[9] H.J. Kee, E.H. Bae, S. Park, K.E. Lee, S.H. Suh, S.W. Kim, M.H. Jeong, HDAC
pathies, and teratogenic effects. Some preexisting pathologies, including
inhibition suppresses cardiac hypertrophy and fibrosis in DOCA-salt hypertensive
coagulopathy or hyperhomocysteinemia and pregnancy, are common rats via regulation of HDAC6/HDAC8 enzyme activity, Kidney Blood Press. Res.
contraindications to VPA usage as a treatment. Balancing therapeutic 37 (4–5) (2013) 229–239.
potentialities with extreme side effects observed following usage of VPA [10] J. Bruni, B.J. Wilder, Valproic acid: review of a new antiepileptic drug, Arch.
Neurol. 36 (7) (1979) 393–398.
to favor the beneficial effects will undoubtedly prove to be a complex [11] B.W. Abou-Khalil, Valproate efficacy in absence seizures is hard to beat:
and challenging problem for medical management. lamotrigine comes close, Epilepsy Curr. 5 (2) (2005) 57–58.
[12] N.C. Spitzer, How GABA generates depolarization, J. Physiol. 588 (Pt 5) (2010)
757–758.
15. Future prospective [13] B. Monti, E. Polazzi, A. Contestabile, Biochemical, molecular and epigenetic
mechanisms of valproic acid neuroprotection, Curr. Mol. Pharmacol. 2 (1) (2009)
From all the previously stated effects, VPA seems to be a pleiotropic 95–109.
[14] C.A. Bradbury, F.L. Khanim, R. Hayden, C.M. Bunce, D.A. White, M.T. Drayson,
agent with multiple mechanisms of action and protective effects against C. Craddock, B.M. Turner, Histone deacetylases in acute myeloid leukaemia show
a number of disease models. Inflammation and oxidative stress are the a distinctive pattern of expression that changes selectively in response to
main pathways in almost all human diseases, so agents possessing the deacetylase inhibitors, Leukemia 19 (10) (2005) 1751–1759.
[15] R.R. Rosato, J.A. Almenara, S. Grant, The histone deacetylase inhibitor MS-275
ability to counteract these pathways are expected to have beneficial promotes differentiation or apoptosis in human leukemia cells through a process
effects in many disease states. VPA was proved to exert these effects, regulated by generation of reactive oxygen species and induction of p21CIP1/
which is why we found these diverse pharmacological activities. WAF1 1, Cancer Res. 63 (13) (2003) 3637–3645.
[16] S. Khan, G. Jena, K. Tikoo, V. Kumar, Valproate attenuates the proteinuria,
Recent reviews suggested the beneficial effect of VPA in SARS-Cov-2
podocyte and renal injury by facilitating autophagy and inactivation of NF-κB/
viral infection based on its anti-thrombotic and anti-inflammatory ef­ iNOS signaling in diabetic rat, Biochimie 110 (2015) 1–16.
fects [112–114]. An in vitro study using human umbilical veins and [17] I. Mannaerts, N.R. Nuytten, V. Rogiers, K. Vanderkerken, L.A. van Grunsven,
coronary artery endothelial cell culture showed that VPA treatment A. Geerts, Chronic administration of valproic acid inhibits activation of mouse
hepatic stellate cells in vitro and in vivo, Hepatology 51 (2) (2010) 603–614.
reduced ACE-2 expression in endothelial cells and the expression of in­ [18] J.L. Hannan, O. Kutlu, B.L. Stopak, X. Liu, F. Castiglione, P. Hedlund, A.
flammatory cytokines IL6 along with the endothelial activation marker L. Burnett, T.J. Bivalacqua, Valproic acid prevents penile fibrosis and erectile

7
D. Singh et al. Biomedicine & Pharmacotherapy 142 (2021) 112021

dysfunction in cavernous nerve-injured rats, J. Sex. Med. 11 (6) (2014) [46] Q. Liu, H. Li, J. Yang, X. Niu, C. Zhao, L. Zhao, Z. Wang, Valproic acid attenuates
1442–1451. inflammation of optic nerve and apoptosis of retinal ganglion cells in a rat model
[19] B. Jin, K.D. Robertson, DNA methyltransferases, DNA damage repair, and cancer, of optic neuritis, Biomed. Pharmacother. 96 (2017) 1363–1370.
Adv. Exp. Med. Biol. 754 (2013) 3–29. [47] J.C. Ximenes, D. de Oliveira Gonçalves, R.M. Siqueira, K.R. Neves, G. Santos
[20] S. Milutinovic, A.C. D’Alessio, N. Detich, M. Szyf, Valproate induces widespread Cerqueira, A.O. Correia, F.H. Félix, L.K. Leal, G.A. de Castro Brito, M. da Graça
epigenetic reprogramming which involves demethylation of specific genes, Naffah-Mazzacorati, G.S. Viana, Valproic acid: an anticonvulsant drug with
Carcinogenesis 28 (3) (2007) 560–571. potent antinociceptive and anti-inflammatory properties, Naunyn
[21] E.N. Olson, J. Backs, T.A. McKinsey (Eds.), Control of cardiac hypertrophy and Schmiedeberg’s Arch. Pharmacol. 386 (7) (2013) 575–587.
heart failure by histone acetylation/deacetylation. Novartis Foundation [48] F. Peña-Ortega, Brain arrhythmias induced by amyloid beta and inflammation:
Symposium, 2006, Wiley Online Library. Involvement in Alzheimer’s disease and other inflammation-related pathologies,
[22] R.-f Li, S.-s Cao, W.-j Fang, Y. Song, X.-T. Luo, H.-y Wang, J.G. Wang, Roles of Curr. Alzheimer Res. 16 (12) (2019) 1108–1131.
HDAC2 and HDAC8 in cardiac remodeling in renovascular hypertensive rats and [49] S. Sharma, K. Sarathlal, R. Taliyan, Epigenetics in neurodegenerative diseases: the
the effects of valproic acid sodium, Pharmacology 99 (1–2) (2017) 27–39. role of histone deacetylases, CNS Neurol. Disord. Drug Targets Former. Curr. Drug
[23] S.-H. Kang, Y.M. Seok, M.-j Song, H.-A. Lee, T. Kurz, I. Kim, Histone deacetylase Targets CNS Neurol. Disord. 18 (1) (2019) 11–18.
inhibition attenuates cardiac hypertrophy and fibrosis through acetylation of [50] X.S. Liu, M. Chopp, H. Kassis, L.F. Jia, A. Hozeska-Solgot, R.L. Zhang, C. Chen, Y.
mineralocorticoid receptor in spontaneously hypertensive rats, Mol. Pharmacol. S. Cui, Z.G. Zhang, Valproic acid increases white matter repair and neurogenesis
87 (5) (2015) 782–791. after stroke, Neuroscience 220 (2012) 313–321.
[24] Y. Liu, S. Li, Z. Zhang, Z. Lv, H. Jiang, X. Tan, F. Liu, Effects of valproic acid on [51] M. Ren, Y. Leng, M. Jeong, P.R. Leeds, D.M. Chuang, Valproic acid reduces brain
sympathetic activity and left ventricularmyocardial remodelling in rats during damage induced by transient focal cerebral ischemia in rats: potential roles of
pressure overload, Turk. J. Med. Sci. 47 (5) (2017) 1651–1660. histone deacetylase inhibition and heat shock protein induction, J. Neurochem.
[25] B. Scholz, J.S. Schulte, S. Hamer, K. Himmler, F. Pluteanu, M.D. Seidl, J. Stein, 89 (6) (2004) 1358–1367.
E. Wardelmann, E. Hammer, U. Völker, F.U. Müller, HDAC (histone deacetylase) [52] Z. Wang, L.-K. Tsai, J. Munasinghe, Y. Leng, E.B. Fessler, F. Chibane, P. Leeds, D.
inhibitor valproic acid attenuates atrial remodeling and delays the onset of atrial M. Chuang, Chronic valproate treatment enhances postischemic angiogenesis and
fibrillation in mice, Circ. Arrhythm. Electrophysiol. 12 (3) (2019), 007071. promotes functional recovery in a rat model of ischemic stroke, Stroke 43 (9)
[26] S. Tian, I. Lei, W. Gao, L. Liu, Y. Guo, J. Creech, T.J. Herron, S. Xian, P.X. Ma, (2012) 2430–2436.
Y. Eugene Chen, Y. Li, H.B. Alam, Z. Wang, HDAC inhibitor valproic acid protects [53] M.R. Silva, A.O. Correia, G.C.A. Dos Santos, L.L.T. Parente, K.P. de Siqueira, D.G.
heart function through Foxm1 pathway after acute myocardial infarction, S. Lima, J.A. Moura, A.E. da Silva Ribeiro, R.O. Costa, D.L. Lucetti, E. Lucetti,
EBioMedicine 39 (2019) 83–94. K. Neves, G.S. de Barros Viana, Neuroprotective effects of valproic acid on brain
[27] J. Choi, S. Park, T. Kwon, S. Sohn, K. Park, J. Kim, Role of the histone deacetylase ischemia are related to its HDAC and GSK3 inhibitions, Pharmacol. Biochem.
inhibitor valproic acid in high-fat diet-induced hypertension via inhibition of Behav. 167 (2018) 17–28.
HDAC1/angiotensin II axis, Int. J. Obes. 41 (11) (2017) 1702–1709. [54] A. Xuan, D. Long, J. Li, W. Ji, L. Hong, M. Zhang, W. Zhang, Neuroprotective
[28] X. Shi, Y. Liu, D. Zhang, D. Xiao, Valproic acid attenuates sepsis-induced effects of valproic acid following transient global ischemia in rats, Life Sci. 90
myocardial dysfunction in rats by accelerating autophagy through the PTEN/ (11–12) (2012) 463–468.
AKT/mTOR pathway, Life Sci. 232 (2019), 116613. [55] J.C. Ximenes, K.R. Neves, L.K. Leal, M.R. do Carmo, G.A. Brito, G. Naffah-
[29] M. Sousa, J. Bruges-Armas, Monogenic diabetes: genetics and relevance on Mazzacoratti Mda, É.A. Cavalheiro, G.S. Viana, Valproic acid neuroprotection in
diabetes mellitus personalized medicine, Curr. Diabetes Rev. 16 (8) (2020) the 6-OHDA model of Parkinson’s disease is possibly related to its anti-
807–819. inflammatory and HDAC inhibitory properties, J. Neurodegener. Dis. 2015
[30] O. Moser, M.L. Eckstein, D.J. West, N. Goswami, H. Sourij, P. Hofmann, Type 1 (2015), 313702.
diabetes and physical exercise: moving (forward) as an adjuvant therapy, Curr. [56] B. Monti, V. Gatta, F. Piretti, S.S. Raffaelli, M. Virgili, A. Contestabile, Valproic
Pharm. Des. 26 (9) (2020) 946–957. acid is neuroprotective in the rotenone rat model of Parkinson’s disease:
[31] E.P. Sørgjerd, Type 1 diabetes-related autoantibodies in different forms of involvement of α-synuclein, Neurotox. Res. 17 (2) (2010) 130–141.
diabetes, Curr. Diabetes Rev. 15 (3) (2019) 199–204. [57] T. Kim, S. Song, Y. Park, S. Kang, H. Seo, HDAC inhibition by valproic acid
[32] S. Khan, G. Jena, Valproic acid improves glucose homeostasis by increasing beta- induces neuroprotection and improvement of PD-like behaviors in LRRK2
cell proliferation, function, and reducing its apoptosis through HDAC inhibition R1441G transgenic mice, Exp. Neurobiol. 28 (4) (2019) 504–515.
in juvenile diabetic rat, J. Biochem. Mol. Toxicol. 30 (9) (2016) 438–446. [58] J. Muangsab, P. Prommeenate, B. Chetsawang, P. Chonpathompikunlert,
[33] S. Khan, S. Kumar, G. Jena, Valproic acid reduces insulin-resistance, fat W. Sukketsiri, P. Hutamekalin, Protective effect of valproic acid on MPP+-
deposition and FOXO1-mediated gluconeogenesis in type-2 diabetic rat, induced neurotoxicity in dopaminergic SH-SY5Y cells through Cdk5/p35/Erk
Biochimie 125 (2016) 42–52. signaling cascade, Trop. J. Pharm. Res. 18 (2019) 11.
[34] A. Terasmaa, U. Soomets, J. Oflijan, M. Punapart, M. Hansen, V. Matto, [59] Y. Leng, D.M. Chuang, Endogenous alpha-synuclein is induced by valproic acid
K. Ehrlich, A. Must, S. Kõks, E. Vasar, Wfs1 mutation makes mice sensitive to through histone deacetylase inhibition and participates in neuroprotection
insulin-like effect of acute valproic acid and resistant to streptozocin, J. Physiol. against glutamate-induced excitotoxicity, J. Neurosci. Off. J. Soc. Neurosci. 26
Biochem. 67 (3) (2011) 381–390. (28) (2006) 7502–7512.
[35] A.J. Akindele, E. Otuguor, D. Singh, D. Ota, A.S. Benebo, Hypoglycemic, [60] H. Qing, G. He, P.T. Ly, C.J. Fox, M. Staufenbiel, F. Cai, Z. Zhang, S. Wei, X. Sun,
antilipidemic and antioxidant effects of valproic acid in alloxan-induced diabetic C.H. Chen, W. Zhou, K. Wang, W. Song, Valproic acid inhibits Abeta production,
rats, Eur. J. Pharmacol. 762 (2015) 174–183. neuritic plaque formation, and behavioral deficits in Alzheimer’s disease mouse
[36] A. Igunnu AO, I. David, O.S. Ogunsola, R.A. Oyegoke, Valproic acid displays anti- models, J. Exp. Med. 205 (12) (2008) 2781–2789.
diabetic and pro-antioxidant effects in high-fat diet and streptozotocin-induced [61] D. Zádori, A. Geisz, E. Vámos, L. Vécsei, P. Klivényi, Valproate ameliorates the
type 2 diabetic rats, Nig J. Pure Appl. Sci. 32 (1) (2019) 3324–3336. survival and the motor performance in a transgenic mouse model of Huntington’s
[37] V. Liakopoulos, S. Roumeliotis, X. Gorny, E. Dounousi, P.R. Mertens, Oxidative disease, Pharmacol. Biochem. Behav. 94 (1) (2009) 148–153.
stress in hemodialysis patients: a review of the literature, Oxid. Med. Cell. [62] Z. Zheng, Y. Wu, Z. Li, L. Ye, Q. Lu, Y. Zhou, Y. Yuan, T. Jiang, L. Xie, Y. Liu,
Longev. 2017 (2017), 3081856. D. Chen, J. Ye, W. Nimlamool, H. Zhang, J. Xiao, Valproic acid affects neuronal
[38] J. Himmelfarb, P. Stenvinkel, T.A. Ikizler, R.M. Hakim, The elephant in uremia: fate and microglial function via enhancing autophagic flux in mice after
oxidant stress as a unifying concept of cardiovascular disease in uremia, Kidney traumatic brain injury, J. Neurochem. 154 (3) (2020) 284–300.
Int. 62 (5) (2002) 1524–1538. [63] C. Zhang, J. Zhu, J. Zhang, H. Li, Z. Zhao, Y. Liao, X. Wang, J. Su, S. Sang,
[39] X.Y. Sun, H.J. Qin, Z. Zhang, Y. Xu, X.C. Yang, D.M. Zhao, X.N. Li, L.K. Sun, X. Yuan, Q. Liu, Neuroprotective and anti-apoptotic effects of valproic acid on
Valproate attenuates diabetic nephropathy through inhibition of endoplasmic adult rat cerebral cortex through ERK and Akt signaling pathway at acute phase
reticulum stress-induced apoptosis, Mol. Med. Rep. 13 (2016) 661–668. of traumatic brain injury, Brain Res. 1555 (2014) 1–9.
[40] K. Van Beneden, C. Geers, M. Pauwels, I. Mannaerts, D. Verbeelen, L.A. van [64] G.M. Hernandez de, F.J. Garay, N.E. Loureiro, Neuroprotective action of valproic
Grunsven, C. Van den Branden, Valproic acid attenuates proteinuria and kidney acid accompanied of the modification on the expression of Bcl-2 and activated
injury, J. Am. Soc. Nephrol. 22 (10) (2011) 1863–1875. caspase-3 in the brain of rats submitted to ischemia/reperfusion, Investig. Clin. 56
[41] Q. Zheng, W. Liu, Z. Liu, H. Zhao, X. Han, M. Zhao, Valproic acid protects septic (4) (2015) 377–388.
mice from renal injury by reducing the inflammatory response, J. Surg. Res. 192 [65] H.S. Go, J.E. Seo, K.C. Kim, S.M. Han, P. Kim, Y.S. Kang, S.H. Han, C.Y. Shin, K.
(1) (2014) 163–169. H. Ko, Valproic acid inhibits neural progenitor cell death by activation of NF-κB
[42] S.L. Berger, The complex language of chromatin regulation during transcription, signaling pathway and up-regulation of Bcl-XL, J. Biomed. Sci. 18 (1) (2011) 48.
Nature 447 (7143) (2007) 407–412. [66] W. Renthal, E.J. Nestler, Histone acetylation in drug addiction, Semin. Cell Dev.
[43] A. Villagra, E.M. Sotomayor, E. Seto, Histone deacetylases and the immunological Biol. (2009).
network: implications in cancer and inflammation, Oncogene 29 (2) (2010) [67] J.-X. Li, R. Han, Y.-P. Deng, S.-Q. Chen, J.-H. Liang, Different effects of valproate
157–173. on methamphetamine-and cocaine-induced behavioral sensitization in mice,
[44] M.A. Amirzargar, F. Yaghubi, M. Hosseinipanah, M. Jafari, M. Pourjafar, Behav. Brain Res. 161 (1) (2005) 125–132.
M. Rezaeepoor, H. Rezaei, G. Roshanaei, M. Hajilooi, G. Solgi, Anti-inflammatory [68] Ø. Kristensen, T. Lølandsmo, Å. Isaksen, J.-K. Vederhus, T. Clausen, Treatment of
effects of valproic acid in a rat model of renal ischemia/reperfusion injury: polydrug-using opiate dependents during withdrawal: towards a standardisation
alteration in cytokine profile, Inflammation 40 (4) (2017) 1310–1318. of treatment, BMC Psychiatry 6 (1) (2006) 1–9.
[45] L.-F. Seet, L.Z. Toh, S.N. Finger, S.W. Chu, T.T. Wong, Valproic acid exerts specific [69] W.H. Organization, Antimicrobial Resistance: Global Report on Surveillance,
cellular and molecular anti-inflammatory effects in post-operative conjunctiva, World Health Organization, 2014.
J. Mol. Med. 97 (1) (2019) 63–75. [70] S.B. Levy, B. Marshall, Antibacterial resistance worldwide: causes, challenges and
responses, Nat. Med. 10 (12) (2004) S122–S129.

8
D. Singh et al. Biomedicine & Pharmacotherapy 142 (2021) 112021

[71] J. Chaillot, F. Tebbji, C. García, H. Wurtele, R. Pelletier, A. Sellam, pH-dependant [93] S. Fushida, J. Kinoshita, M. Kaji, K. Oyama, Y. Hirono, T. Tsukada, T. Fujimura,
antifungal activity of valproic acid against the human fungal pathogen Candida T. Ohta, Paclitaxel plus valproic acid versus paclitaxel alone as second-or third-
albicans, Front. Microbiol. 8 (2017) 1956. line therapy for advanced gastric cancer: a randomized Phase II trial, Drug Des.
[72] R.N. Mahalingam G, I. Purushothaman, A V, K S, Evaluation of in vitro Dev. Ther. 10 (2016) 2353–2358.
antibacterial activity of Anticonvulsant drugs, J. Pharma Res. 4 (1) (2015) 13–15. [94] Z. Xie, Y. Ago, N. Okada, M. Tachibana, Valproic acid attenuates
[73] D.M. Vigushin, R.C. Coombes, Histone deacetylase inhibitors in cancer treatment, immunosuppressive function of myeloid-derived suppressor cells, J. Pharmacol.
Anti Cancer Drugs 13 (1) (2002) 1–13. Sci. 137 (4) (2018) 359–365.
[74] Y. Bai, D. Ahmad, T. Wang, G. Cui, W. Li, Research advances in the use of histone [95] I. Dalkilic, L.M. Kunkel, Muscular dystrophies: genes to pathogenesis, Curr. Opin.
deacetylase inhibitors for epigenetic targeting of cancer, Curr. Top. Med. Chem. Genet. Dev. 13 (3) (2003) 231–238.
19 (12) (2019) 995–1004. [96] T.A. McKinsey, C.L. Zhang, E.N. Olson, Activation of the myocyte enhancer
[75] S. Jawed, B. Kim, T. Ottenhof, G.M. Brown, E.S. Werstiuk, L.P. Niles, Human factor-2 transcription factor bycalcium/calmodulin-dependent protein kinase-
melatonin MT1 receptor induction by valproic acid and its effects in combination stimulated binding of 14–3-3 tohistone deacetylase 5, Proc. Natl. Acad. Sci. USA
with melatonin on MCF-7 breast cancer cell proliferation, Eur. J. Pharmacol. 560 97 (26) (2000) 14400–14405.
(1) (2007) 17–22. [97] P.B. Gurpur, J. Liu, D.J. Burkin, S.J. Kaufman, Valproic acid activates the PI3K/
[76] M. Terranova-Barberio, M.S. Roca, A.I. Zotti, A. Leone, F. Bruzzese, C. Vitagliano, Akt/mTOR pathway in muscle and ameliorates pathology in a mouse model of
G. Scogliamiglio, D. Russo, G. D’Angelo, R. Franco, A. Budillon, E. Di Gennaro, Duchenne muscular dystrophy, Am. J. Pathol. 174 (3) (2009) 999–1008.
Valproic acid potentiates the anticancer activity of capecitabine in vitro and in [98] D. Palacios, P.L. Puri, The epigenetic network regulating muscle development and
vivo in breast cancer models via induction of thymidine phosphorylase regeneration, J. Cell. Physiol. 207 (1) (2006) 1–11.
expression, Oncotarget 7 (7) (2016) 7715–7731. [99] R. Sun, S. Zhang, W. Hu, X. Lu, N. Lou, Z. Yang, S. Chen, X. Zhang, H. Yang,
[77] H.K. Park, B.R. Han, W.H. Park, Combination of arsenic trioxide and valproic acid Valproic acid attenuates skeletal muscle wasting by inhibiting C/EBPβ-regulated
efficiently inhibits growth of lung cancer cells via G2/M-phase arrest and atrogin1 expression in cancer cachexia, Am. J. Physiol. Cell Physiol. 311 (1)
apoptotic cell death, Int. J. Mol. Sci. 21 (7) (2020) 2649. (2016) C101–C115.
[78] S. Li, J.K. Zhan, Y.J. Wang, X. Lin, J.Y. Zhong, Y. Wang, P. Tan, J.Y. He, X.J. Cui, [100] K.J. Swoboda, C.B. Scott, S.P. Reyna, T.W. Prior, B. LaSalle, S.L. Sorenson,
Y.Y. Chen, W. Huang, Y.S. Liu, Combination chemotherapy of valproic acid (VPA) J. Wood, G. Acsadi, T.O. Crawford, J.T. Kissel, K.J. Krosschell, G. D’Anjou, M.
and gemcitabine regulates STAT3/Bmi1 pathway to differentially potentiate the B. Bromberg, M.K. Schroth, G.M. Chan, B. Elsheikh, L.R. Simard, Phase II open
motility of pancreatic cancer cells, Cell Biosci. 9 (1) (2019) 1–15. label study of valproic acid in spinal muscular atrophy, PLoS One 4 (5) (2009),
[79] N. Shirsath, M. Rathos, U. Chaudhari, H. Sivaramakrishnan, K. Joshi, Potentiation 5268.
of anticancer effect of valproic acid, an antiepileptic agent with histone [101] Z. Zhang, N. Tong, Y. Gong, Q. Qiu, L. Yin, X. Lv, X. Wu, Valproate protects the
deacetylase inhibitory activity, by the cyclin-dependent kinase inhibitor P276-00 retina from endoplasmic reticulum stress-induced apoptosis after ischemia-
in human non-small-cell lung cancer cell lines, Lung Cancer 82 (2) (2013) reperfusion injury, Neurosci. Lett. 504 (2) (2011) 88–92.
214–221. [102] Z. Zhang, X. Qin, N. Tong, X. Zhao, Y. Gong, Y. Shi, X. Wu, Valproic acid-mediated
[80] A. Phillips, T. Bullock, N. Plant, Sodium valproate induces apoptosis in the rat neuroprotection in retinal ischemia injury via histone deacetylase inhibition and
hepatoma cell line, FaO, Toxicology 192 (2–3) (2003) 219–227. transcriptional activation, Exp. Eye Res. 94 (1) (2012) 98–108.
[81] T. Lin, Q. Ren, W. Zuo, R. Jia, L. Xie, R. Lin, H. Zhao, J. Chen, Y. Lei, P. Wang, [103] Z. Zhang, X. Qin, X. Zhao, N. Tong, Y. Gong, W. Zhang, X. Wu, Valproic acid
H. Dong, L. Huang, J. Cai, Y. Peng, Z. Yu, J. Tan, S. Wang, Valproic acid exhibits regulates antioxidant enzymes and prevents ischemia/reperfusion injury in the
anti-tumor activity selectively against EGFR/ErbB2/ErbB3-coexpressing rat retina, Curr. Eye Res. 37 (5) (2012) 429–437.
pancreatic cancer via induction of ErbB family members-targeting microRNAs, [104] K. Kim, Y. Li, G. Jin, W. Chong, B. Liu, J. Lu, K. Lee, M. Demoya, G.C. Velmahos,
J. Exp. Clin. Cancer Res. 38 (1) (2019) 1–12. H.B. Alam, Effect of valproic acid on acute lung injury in a rodent model of
[82] M. Wei, S. Mao, G. Lu, L. Li, X. Lan, Z. Huang, Y. Chen, M. Zhao, Y. Zhao, Q. Xia, intestinal ischemia reperfusion, Resuscitation 83 (2) (2012) 243–248.
Valproic acid sensitizes metformin-resistant human renal cell carcinoma cells by [105] S.Y. Wu, S.E. Tang, F.C. Ko, G.C. Wu, K.L. Huang, S.J. Chu, Valproic acid
upregulating H3 acetylation and EMT reversal, BMC Cancer 18 (1) (2018) 1–15. attenuates acute lung injury induced by ischemia-reperfusion in rats,
[83] D.Y. Greenblatt, M.A. Cayo, J.T. Adler, L. Ning, M.R. Haymart, Anesthesiology 122 (6) (2015) 1327–1337.
M. Kunnimalaiyaan, H. Chen, Valproic acid activates Notch1 signaling and [106] A.W. Beharry, P.B. Sandesara, B.M. Roberts, L.F. Ferreira, S.M. Senf, A.R. Judge,
induces apoptosis in medullary thyroid cancer cells, Ann. Surg. 247 (6) (2008) HDAC1 activates FoxO and is both sufficient and required for skeletal muscle
1036–1040. atrophy, J. Cell Sci. 127 (Pt 7) (2014) 1441–1453.
[84] M. Michaelis, T. Suhan, J. Cinatl, P.H. Driever, J. Cinatl, Valproic acid and [107] Y.C. Tseng, S.K. Kulp, I.L. Lai, E.C. Hsu, W.A. He, D.E. Frankhouser, P.S. Yan,
interferon-α synergistically inhibit neuroblastoma cell growth in vitro and in vivo, X. Mo, M. Bloomston, G.B. Lesinski, G. Marcucci, D.C. Guttridge, T. Bekaii-Saab,
Int. J. Oncol. 25 (6) (2004) 1795–1799. C.S. Chen, Preclinical investigation of the novel histone deacetylase inhibitor AR-
[85] J. Sun, J. Piao, N. Li, Y. Yang, K.Y. Kim, Z. Lin, Valproic acid targets HDAC1/2 42 in the treatment of cancer-induced cachexia, J. Natl. Cancer Inst. 107 (12)
and HDAC1/PTEN/Akt signalling to inhibit cell proliferation via the induction of (2015), 274.
autophagy in gastric cancer, FEBS J. 287 (10) (2020) 2118–2133. [108] F. Penna, P. Costelli, New developments in investigational HDAC inhibitors for
[86] W. Li, L. Ma, Synergistic antitumor activity of oridonin and valproic acid on HL- the potential multimodal treatment of cachexia, Expert Opin. Investig. Drugs 28
60 leukemia cells, J. Cell. Biochem. 120 (4) (2019) 5620–5627. (2) (2019) 179–189.
[87] Z. Zhang, R. Zhang, C. Hao, X. Pei, J. Li, L. Wang, GANT61 and valproic acid [109] D.C. Lagace, R.S. McLeod, M.W. Nachtigal, Valproic acid inhibits leptin secretion
synergistically inhibited multiple myeloma cell proliferation via Hedgehog and reduces leptin messenger ribonucleic acid levels in adipocytes, Endocrinology
signaling pathway, Med. Sci. Monit. Int. Med. J. Exp. Clin. Res. 26 (2020) 145 (12) (2004) 5493–5503.
e920541-e920541. [110] M.A. Elbadawi, M.K. Awadalla, M.M. Hamid, M.A. Mohamed, T.A. Awad,
[88] A.I. Daud, J. Dawson, R.C. DeConti, E. Bicaku, D. Marchion, S. Bastien, F. Valproic acid as a potential inhibitor of Plasmodium falciparum histone
A. Hausheer, R. Lush, A. Neuger, D.M. Sullivan, P.N. Munster, Potentiation of a deacetylase 1 (PfHDAC1): an in silico approach, Int. J. Mol. Sci. 16 (2) (2015)
topoisomerase I inhibitor, karenitecin, by the histone deacetylase inhibitor 3915–3931.
valproic acid in melanoma: translational and phase I/II clinical trial, Clin. Cancer [111] S. Piplani, P.K. Verma, A. Kumar, Neuroinformatics analyses reveal GABAt and
Res. 15 (7) (2009) 2479–2487. SSADH as major proteins involved in anticonvulsant activity of valproic acid,
[89] A. Rocca, S. Minucci, G. Tosti, D. Croci, F. Contegno, M. Ballarini, F. Nolè, Biomed. Pharmacother. Biomed. Pharmacother. 81 (2016) 402–410.
E. Munzone, A. Salmaggi, A. Goldhirsch, P.G. Pelicci, A. Testori, A phase I–II [112] P. Bhargava, P. Panda, V. Ostwal, A. Ramaswamy, Repurposing valproate to
study of the histone deacetylase inhibitor valproic acid plus prevent acute respiratory distress syndrome/acute lung injury in COVID-19: a
chemoimmunotherapy in patients with advanced melanoma, Br. J. Cancer 100 review of immunomodulatory action, Cancer Res. Stat. Treat. 3 (5) (2020) 65–70.
(1) (2009) 28–36. [113] G. Unal, B. Turan, Y.H. Balcioglu, Immunopharmacological management of
[90] H. Fredly, E. Ersvær, A.O. Kittang, G. Tsykunova, B.T. Gjertsen, Ø. Bruserud, The COVID-19: potential therapeutic role of valproic acid, Med. Hypotheses 143
combination of valproic acid, all-trans retinoic acid and low-dose cytarabine as (2020), 109891.
disease-stabilizing treatment in acute myeloid leukemia, Clin. Epigenetics 5 (1) [114] B. Pitt, N.R. Sutton, Z. Wang, S.N. Goonewardena, M. Holinstat, Potential
(2013) 1–15. repurposing of the HDAC inhibitor valproic acid for patients with COVID-19, Eur.
[91] D.W. Coulter, C. Walko, J. Patel, B.M. Moats-Staats, A. McFadden, S.V. Smith, W. J. Pharmacol. 898 (2021), 173988.
A. Khan, A.S. Bridges, A.M. Deal, J. Oesterheld, I.J. Davis, J. Blatt, Valproic acid [115] S. Singh, K. Singh, Valproic Acid in Prevention and Treatment of COVID-19,
reduces the tolerability of temsirolimus in children and adolescents with solid Authorea Preprints, 2020.
tumors, Anti Cancer Drugs 24 (4) (2013) 415–421. [116] E. Chiquete, L. Toapanta-Yanchapaxi, C. Cantu-Brito, Methods of an open-label
[92] J.M. Su, X.-N. Li, P. Thompson, C.-N. Ou, A.M. Ingle, H. Russell, C.C. Lau, P. proof-of-concept trial of intravenous valproic acid for severe COVID-19, medRxiv,
C. Adamson, S.M. Blaney, Phase 1 study of valproic acid in pediatric patients with 2020.
refractory solid or CNS tumors: a children’s oncology group report, Clin. Cancer
Res. 17 (3) (2011) 589–597.

You might also like